1
|
Ramar V, Guo S, Hudson B, Liu M. Progress in Glioma Stem Cell Research. Cancers (Basel) 2023; 16:102. [PMID: 38201528 PMCID: PMC10778204 DOI: 10.3390/cancers16010102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 12/15/2023] [Accepted: 12/22/2023] [Indexed: 01/12/2024] Open
Abstract
Glioblastoma multiforme (GBM) represents a diverse spectrum of primary tumors notorious for their resistance to established therapeutic modalities. Despite aggressive interventions like surgery, radiation, and chemotherapy, these tumors, due to factors such as the blood-brain barrier, tumor heterogeneity, glioma stem cells (GSCs), drug efflux pumps, and DNA damage repair mechanisms, persist beyond complete isolation, resulting in dismal outcomes for glioma patients. Presently, the standard initial approach comprises surgical excision followed by concurrent chemotherapy, where temozolomide (TMZ) serves as the foremost option in managing GBM patients. Subsequent adjuvant chemotherapy follows this regimen. Emerging therapeutic approaches encompass immunotherapy, including checkpoint inhibitors, and targeted treatments, such as bevacizumab, aiming to exploit vulnerabilities within GBM cells. Nevertheless, there exists a pressing imperative to devise innovative strategies for both diagnosing and treating GBM. This review emphasizes the current knowledge of GSC biology, molecular mechanisms, and associations with various signals and/or pathways, such as the epidermal growth factor receptor, PI3K/AKT/mTOR, HGFR/c-MET, NF-κB, Wnt, Notch, and STAT3 pathways. Metabolic reprogramming in GSCs has also been reported with the prominent activation of the glycolytic pathway, comprising aldehyde dehydrogenase family genes. We also discuss potential therapeutic approaches to GSC targets and currently used inhibitors, as well as their mode of action on GSC targets.
Collapse
Affiliation(s)
- Vanajothi Ramar
- Department of Microbiology, Biochemistry & Immunology, Morehouse School of Medicine, Atlanta, GA 30310, USA; (V.R.); (B.H.)
| | - Shanchun Guo
- Department of Chemistry, Xavier University, 1 Drexel Dr., New Orleans, LA 70125, USA;
| | - BreAnna Hudson
- Department of Microbiology, Biochemistry & Immunology, Morehouse School of Medicine, Atlanta, GA 30310, USA; (V.R.); (B.H.)
| | - Mingli Liu
- Department of Microbiology, Biochemistry & Immunology, Morehouse School of Medicine, Atlanta, GA 30310, USA; (V.R.); (B.H.)
| |
Collapse
|
2
|
Salari N, Rasoulpoor S, Shabani S, Mansouri K, Bokaee S, Fatahian R, Farshchian N, Mohammadi M, Hosseinian-Far M. ERCC2 rs13181 Polymorphism Association with Glioma Risk: an Update Meta-Analysis. Indian J Surg Oncol 2023; 14:60-68. [PMID: 36891435 PMCID: PMC9986186 DOI: 10.1007/s13193-022-01623-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Accepted: 08/10/2022] [Indexed: 10/15/2022] Open
Abstract
Glioma is the most common type of primary brain tumour which accounts for about 30% of all brain and central nervous system tumours, and approximately 70% of adult malignant brain tumours. Numerous studies have been performed to assess the relationship between ERCC2 rs13181 polymorphism and the risk of glioma development, yet these findings of these studies are often inconsistent and contradictory. Therefore, the aim of this study is to conduct a systematic review and meta-analysis to assess the role of ERCC2 rs13181 in glioma developing. In this work, we have conducted a systematic review and meta-analysis. In order to collect the results of relevant studies on the association of ERCC2 rs13181 gene polymorphism with glioma, we initially searched the Scopus, Embase, Web of Science (WoS), PubMed, and ScienceDirect databases, without a lower time limit, and until June 2020. In order to analyse the eligible studies, the random effects model was used and the heterogeneity of the studies was investigated with the I 2 index. Data analysis was performed within the Comprehensive Meta-Analysis software (version 2). The total number of studies that focused on patients with glioma was 10. The odds ratio of GG vs TT genotype in patients with glioma based on meta-analysis was 1.08 (0.85-1.37: 95% confidence interval), which indicates the increasing effect of GG vs TT genotype by 0.08. The odds ratio of GG + TG vs TT genotype in patients with glioma was 1.22 (1.38-1.7: 95% confidence interval) based on meta-analysis, which indicates the increasing effect of GG + TG vs TT genotype as 0.22. The odds ratio of TG vs TT genotype in patients with glioma was 1.2 (0.38-1.4: 95% confidence interval), which shows the increasing effect of TG vs TT genotype by 0.2. The odds ratio of G vs T genotype in patients with glioma based on the meta-analysis was 1.15 (1.26-1.4: 95% confidence interval), which indicates the increasing effect of G vs T genotype by 0.15. The odds ratio of GG vs TG + TT genotype in patients with glioma based on meta-analysis was 1.22 (1.33-1.45: 95% confidence interval), which indicates the increasing effect of GG vs TG + TT genotype by 0.22. The results of this systematic review and meta-analysis show that ERCC2 rs13181 polymorphism and its genotypes are an important risk factor for genetic susceptibility to glioma tumour.
Collapse
Affiliation(s)
- Nader Salari
- Department of Biostatistics, School of Health, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Shna Rasoulpoor
- Student Research Committee, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Shervin Shabani
- Student Research Committee, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Kamran Mansouri
- Medical Biology Research Centre, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Shadi Bokaee
- Faculty of Health and Life Sciences, School of Life Sciences, Coventry University, Coventry, UK
| | - Reza Fatahian
- Department of Neurosurgery, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Negin Farshchian
- Department of Radiation Oncology, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Masoud Mohammadi
- Cellular and Molecular Research Center, Gerash University of Medical Sciences, Gerash, Iran
| | - Melika Hosseinian-Far
- Department of Food Science & Technology, Faculty of Agriculture, Ferdowsi University of Mashhad (FUM), Mashhad, Iran
| |
Collapse
|
3
|
Association between XRCC3 p.Thr241Met polymorphism and risk of glioma: A systematic review and meta-analysis. PLoS One 2022; 17:e0276313. [PMID: 36264998 PMCID: PMC9584405 DOI: 10.1371/journal.pone.0276313] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Accepted: 10/04/2022] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND The XRCC3 p.Thr241Met (rs861539) polymorphism has been extensively studied for its association with glioma risk, but results remain conflicting. Therefore, we performed a systematic review and meta-analysis to resolve this inconsistency. METHODS Studies published up to June 10, 2022, were searched in PubMed, Web of Science, Scopus, VIP, Wanfang, and China National Knowledge Infrastructure databases and screened for eligibility. Then, the combined odds ratio (OR) of the included studies was estimated based on five genetic models, i.e., homozygous (Met/Met vs. Thr/Thr), heterozygous (Thr/Met vs. Thr/Thr), dominant (Thr/Met + Met/Met vs. Thr/Thr), recessive (Met/Met vs. Thr/Thr + Thr/Met) and allele (Met vs. Thr). The study protocol was preregistered at PROSPERO (registration number: CRD42021235704). RESULTS Overall, our meta-analysis of 14 eligible studies involving 12,905 subjects showed that the p.Thr241Met polymorphism was significantly associated with increased glioma risk in both homozygous and recessive models (homozygous, OR = 1.381, 95% CI = 1.081-1.764, P = 0.010; recessive, OR = 1.305, 95% CI = 1.140-1.493, P<0.001). Subgroup analyses by ethnicity also revealed a statistically significant association under the two aforementioned genetic models, but only in the Asian population and not in Caucasians (P>0.05). CONCLUSION We demonstrated that the XRCC3 p.Thr241Met polymorphism is associated with an increased risk of glioma only in the homozygous and recessive models.
Collapse
|
4
|
Maksoud S. The DNA Double-Strand Break Repair in Glioma: Molecular Players and Therapeutic Strategies. Mol Neurobiol 2022; 59:5326-5365. [PMID: 35696013 DOI: 10.1007/s12035-022-02915-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Accepted: 06/05/2022] [Indexed: 12/12/2022]
Abstract
Gliomas are the most frequent type of tumor in the central nervous system, which exhibit properties that make their treatment difficult, such as cellular infiltration, heterogeneity, and the presence of stem-like cells responsible for tumor recurrence. The response of this type of tumor to chemoradiotherapy is poor, possibly due to a higher repair activity of the genetic material, among other causes. The DNA double-strand breaks are an important type of lesion to the genetic material, which have the potential to trigger processes of cell death or cause gene aberrations that could promote tumorigenesis. This review describes how the different cellular elements regulate the formation of DNA double-strand breaks and their repair in gliomas, discussing the therapeutic potential of the induction of this type of lesion and the suppression of its repair as a control mechanism of brain tumorigenesis.
Collapse
Affiliation(s)
- Semer Maksoud
- Experimental Therapeutics and Molecular Imaging Unit, Department of Neurology, Neuro-Oncology Division, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA.
| |
Collapse
|
5
|
ÇAĞLAR HO. Identification of Genes Related to DNA Repair Mechanism in Glioblastoma by Bioinformatics Methods. KOCAELI ÜNIVERSITESI SAĞLIK BILIMLERI DERGISI 2022. [DOI: 10.30934/kusbed.1003777] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Objective: Aberrant expression of genes involved in DNA repair mechanisms (DRM) have been associated with radiation sensitivity of glioblastoma (GBM) cells. Identification of genes in DRM through bioinformatics methods may help identify potential novel therapeutic targets that can be used in GBM treatment. This study aims to identify genes that play a role in DRM in GBM using bioinformatics methods.
Methods: Genes associated with DRM were identified using the “Reactome” and “KEGG” databases. The mRNA expression profiles of DRM related genes were analyzed in the GEO GDS1813 and GDS2853 datasets including GBM tumor samples using the "Orange Canvas" software. Genetic changes of genes were identified in GBM TCGA cases using the cBioPortal database. The GEPIA2 was used to show the effect of altered expression profiles of these genes on patient survival.
Results: The mRNA expression profiles of ERCC6, FAN1, MBD4, PARP1 and UNG genes were found to be altered in GBM tumors. Mutations and copy number alterations for the identified genes were observed in TCGA GBM cases. The overall survival and disease-free survival of TCGA GBM patients were not significantly different between high and low expression groups.
Conclusion: ERCC6, PARP1 and UNG genes identified in the current study may be potential therapeutic targets that can increase the efficacy of radiotherapy in GBM in case of their suppression.
Collapse
|
6
|
Bueno-Martínez E, Lara-Almunia M, Rodríguez-Arias C, Otero-Rodríguez A, Garfias-Arjona S, González-Sarmiento R. Polymorphisms in autophagy genes are genetic susceptibility factors in glioblastoma development. BMC Cancer 2022; 22:146. [PMID: 35123435 PMCID: PMC8818195 DOI: 10.1186/s12885-022-09214-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Accepted: 01/19/2022] [Indexed: 12/31/2022] Open
Abstract
Background Glioblastoma is the most aggressive and common malignant primary brain tumor in adults. Many genetic, epigenetic and genomic mutations have been identified in this tumor, but no driving cause has been identified yet for glioblastoma pathogenesis. Autophagy has proved to be deregulated in different diseases such as cancer where it has a dual role, acting as a tumor suppression mechanism during the first steps of tumor development and promoting cancer cells survival in stablished tumors. Methods Here, we aimed to assess the potential association between several candidate polymorphisms in autophagy genes (ATG2B rs3759601, ATG16L1 rs2241880, ATG10 rs1864183, ATG5 rs2245214, NOD2 rs2066844 and rs2066845) and glioblastoma susceptibility. Results Our results showed a significant correlation between ATG2B rs3759601, ATG10 rs1864183 and NOD2 rs2066844 variants and higher risk to suffer glioblastoma. In addition, the relationship between the different clinical features listed in glioblastoma patients and candidate gene polymorphisms was also investigated, finding that ATG10 rs1864183 might be a promising prognosis factor for this tumor. Conclusions This is the first report evaluating the role of different variants in autophagy genes in modulating glioblastoma risk and our results emphasize the importance of autophagy in glioblastoma development. Supplementary Information The online version contains supplementary material available at 10.1186/s12885-022-09214-y.
Collapse
|
7
|
Di Cintio F, Dal Bo M, Baboci L, De Mattia E, Polano M, Toffoli G. The Molecular and Microenvironmental Landscape of Glioblastomas: Implications for the Novel Treatment Choices. Front Neurosci 2020; 14:603647. [PMID: 33324155 PMCID: PMC7724040 DOI: 10.3389/fnins.2020.603647] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Accepted: 11/03/2020] [Indexed: 12/20/2022] Open
Abstract
Glioblastoma (GBM) is the most frequent and aggressive primary central nervous system tumor. Surgery followed by radiotherapy and chemotherapy with alkylating agents constitutes standard first-line treatment of GBM. Complete resection of the GBM tumors is generally not possible given its high invasive features. Although this combination therapy can prolong survival, the prognosis is still poor due to several factors including chemoresistance. In recent years, a comprehensive characterization of the GBM-associated molecular signature has been performed. This has allowed the possibility to introduce a more personalized therapeutic approach for GBM, in which novel targeted therapies, including those employing tyrosine kinase inhibitors (TKIs), could be employed. The GBM tumor microenvironment (TME) exerts a key role in GBM tumor progression, in particular by providing an immunosuppressive state with low numbers of tumor-infiltrating lymphocytes (TILs) and other immune effector cell types that contributes to tumor proliferation and growth. The use of immune checkpoint inhibitors (ICIs) has been successfully introduced in numerous advanced cancers as well as promising results have been shown for the use of these antibodies in untreated brain metastases from melanoma and from non-small cell lung carcinoma (NSCLC). Consequently, the use of PD-1/PD-L1 inhibitors has also been proposed in several clinical trials for the treatment of GBM. In the present review, we will outline the main GBM molecular and TME aspects providing also the grounds for novel targeted therapies and immunotherapies using ICIs for GBM.
Collapse
Affiliation(s)
- Federica Di Cintio
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico di Aviano, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Aviano, Italy
- Department of Life Sciences, University of Trieste, Trieste, Italy
| | - Michele Dal Bo
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico di Aviano, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Aviano, Italy
| | - Lorena Baboci
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico di Aviano, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Aviano, Italy
| | - Elena De Mattia
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico di Aviano, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Aviano, Italy
| | - Maurizio Polano
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico di Aviano, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Aviano, Italy
| | - Giuseppe Toffoli
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico di Aviano, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Aviano, Italy
| |
Collapse
|
8
|
High tumor mutational burden and T-cell activation are associated with long-term response to anti-PD1 therapy in Lynch syndrome recurrent glioblastoma patient. Cancer Immunol Immunother 2020; 70:831-842. [PMID: 33140187 DOI: 10.1007/s00262-020-02769-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Accepted: 10/15/2020] [Indexed: 12/11/2022]
Abstract
BACKGROUND Glioblastomas (GBMs) in patients harboring somatic or germinal mutations of mismatch-repair (MMR) genes exhibit a hypermutable phenotype. Here, we describe a GBM patient with increased tumor mutational burden and germline MMR mutations, treated using anti-PD1 therapy. METHODS A woman with newly diagnosed GBM (nGBM) was treated by surgery, radiotherapy, and temozolomide. The tumor recurred after 13 months leading to a second surgery and treatment with nivolumab. Whole-exome sequencing was performed on the nGBM, recurrent GBM (rGBM), and blood. Immune infiltration was investigated by immunohistochemistry and the immune response in the blood during treatment was analyzed by flow cytometry. RESULTS High density of infiltrating CD163 + cells was found in both GBM specimens. Large numbers of CD3 + and CD8 + T cells were homogeneously distributed in the nGBM. The infiltration of CD4 + T cells and a different CD8 + T cell density were observed in the rGBM. Both GBM shared 12,431 somatic mutations, with 113 substitutions specific to the nGBM and 1,683 specific to the rGBM. Germline variants included pathogenic mutation in the MSH2 (R359S) gene, suggesting the diagnosis of Lynch syndrome. Systemic immunophenotyping revealed the generation of CD8 + T memory cells and persistent activation of CD4 + T cells. The patient is still receiving nivolumab 68 months after the second surgery. CONCLUSIONS Our observations indicate that the hypermutator phenotype associated with germinal mutations of MMR genes and abundant T-cell infiltration contributes to a durable clinical benefit sustained by a persistent and robust immune response during anti-PD1 therapy.
Collapse
|
9
|
DNA Mismatch Repair Gene Variants in Sporadic Solid Cancers. Int J Mol Sci 2020; 21:ijms21155561. [PMID: 32756484 PMCID: PMC7432688 DOI: 10.3390/ijms21155561] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 07/29/2020] [Accepted: 07/30/2020] [Indexed: 12/18/2022] Open
Abstract
The phenotypic effects of single nucleotide polymorphisms (SNPs) in the development of sporadic solid cancers are still scarce. The aim of this review was to summarise and analyse published data on the associations between SNPs in mismatch repair genes and various cancers. The mismatch repair system plays a unique role in the control of the genetic integrity and it is often inactivated (germline and somatic mutations and hypermethylation) in cancer patients. Here, we focused on germline variants in mismatch repair genes and found the outcomes rather controversial: some SNPs are sometimes ascribed as protective, while other studies reported their pathological effects. Regarding the complexity of cancer as one disease, we attempted to ascertain if particular polymorphisms exert the effect in the same direction in the development and treatment of different malignancies, although it is still not straightforward to conclude whether polymorphisms always play a clear positive role or a negative one. Most recent and robust genome-wide studies suggest that risk of cancer is modulated by variants in mismatch repair genes, for example in colorectal cancer. Our study shows that rs1800734 in MLH1 or rs2303428 in MSH2 may influence the development of different malignancies. The lack of functional studies on many DNA mismatch repair SNPs as well as their interactions are not explored yet. Notably, the concerted action of more variants in one individual may be protective or harmful. Further, complex interactions of DNA mismatch repair variations with both the environment and microenvironment in the cancer pathogenesis will deserve further attention.
Collapse
|
10
|
Tavares CB, Gomes-Braga FDCSA, Sousa EB, Brito JNPDO, Melo MDA, Campelo V, Neto FM, de Araújo RML, Kessler IM, Sousa Júnior LDM, Filho LCC, Aguiar YQ, Lopes Costa PV, da Silva BB. Association between Single Nucleotide Polymorphisms and Glioma Risk: A Systematic Literature Review. Cancer Invest 2020; 38:169-183. [PMID: 31957502 DOI: 10.1080/07357907.2020.1719502] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
This study aimed to determine the main single nucleotide polymorphisms (SNPs) that are associated with an increased or decreased risk of glioma development in healthy individuals. We conducted a systematic review of the articles published in English on the PUBMED database between January 2008 and December 2017. Our search resulted in a total of 743 articles; however, only 56 were included in this review. A total of 148 polymorphisms were found, which involved 64 different genes. The polymorphisms that were most associated with an increased risk of glioma development were polymorphic variants rs179782, rs13181, and rs3791679 of the genes XRCC1, ERCC2, and EFEMP1, respectively.
Collapse
Affiliation(s)
- Cléciton Braga Tavares
- Postgraduate Program of the Northeast Network of Biotechnology (RENORBIO), Federal University of Piauí, Teresina, Brazil
| | | | | | | | | | - Viriato Campelo
- Postgraduate Program in Health Sciences, Federal University of Piauí, Teresina, Brazil
| | | | | | | | | | | | | | | | - Benedito Borges da Silva
- Postgraduate Program of the Northeast Network of Biotechnology (RENORBIO), Federal University of Piauí, Teresina, Brazil
| |
Collapse
|
11
|
Shao WH, Wang CY, Wang LY, Xiao F, Xiao DS, Yang H, Long XY, Zhang L, Luo HG, Yin JY, Wu W. A Hereditable Mutation of MSH2 Gene Associated with Lynch Syndrome in a Five Generation Chinese Family. Cancer Manag Res 2020; 12:1469-1482. [PMID: 32161499 PMCID: PMC7051253 DOI: 10.2147/cmar.s222572] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Accepted: 12/24/2019] [Indexed: 12/12/2022] Open
Abstract
Purpose In order to clarify which variants of the MMR gene could provide current "healthy" members in affected families a more accurate risk assessment or predictive testing. Patients and Methods One family, which meets the criteria according to both Amsterdam I/II and Bethesda guidelines, is reported in this study. The proband and some relatives of the patient have been investigated for whole genome sequencing, microsatellite instability, immunohistochemical MMR protein staining and verified by Sanger sequencing. Results A heterozygous insertion of uncertain significance (c.420dup, p.Met141Tyrfs) in MSH2 gene was found in proband (III-16) and part of His relatives. The variant was associated with a lack of expression of MSH2 protein (MMR deficient) and high microsatellite instability analysis (MSI) status in tumor tissues of LS patients. In addition, we found that the variant could affect the expression of MSH2 and the response to chemotherapy drugs in vitro. Conclusion We identified an insertion mutation (rs1114167810, c.420dup, p.Met141Tyrfs) in MSH2 in LS using whole genome-wide sequencing (WGS). We further confirmed that this mutation plays an important role in LS patients of this pedigree based on in vivo and vitro study.
Collapse
Affiliation(s)
- Wei-Hua Shao
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha 410078, People's Republic of China.,Institute of Clinical Pharmacology, Central South University; Hunan Key Laboratory of Pharmacogenetics, Changsha 410078, People's Republic of China.,Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha 410078, People's Republic of China.,Department of Geratic Surgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, People's Republic of China
| | - Cheng-Yu Wang
- Department of Geratic Surgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, People's Republic of China.,National Clinical Research Center for Geriatric Disorders, Changsha, Hunan 410008, People's Republic of China
| | - Lei-Yun Wang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha 410078, People's Republic of China.,Institute of Clinical Pharmacology, Central South University; Hunan Key Laboratory of Pharmacogenetics, Changsha 410078, People's Republic of China.,Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha 410078, People's Republic of China.,Department of Geratic Surgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, People's Republic of China
| | - Fan Xiao
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha 410078, People's Republic of China.,Institute of Clinical Pharmacology, Central South University; Hunan Key Laboratory of Pharmacogenetics, Changsha 410078, People's Republic of China.,Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha 410078, People's Republic of China.,Department of Geratic Surgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, People's Republic of China
| | - De-Sheng Xiao
- Department of Pathology, Xiangya Hospital/School of Basic Medicine, Central South University, Changsha 410078, Hunan, People's Republic of China
| | - Hao Yang
- Department of Geratic Surgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, People's Republic of China.,National Clinical Research Center for Geriatric Disorders, Changsha, Hunan 410008, People's Republic of China
| | - Xue-Ying Long
- Department of Radiology, Xiangya Hospital, Central South University, Changsha 410008, People's Republic of China
| | - Le Zhang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China
| | - Heng-Gui Luo
- Department of General Surgery, The Central Hospital of Xiangtan City, Xiangtan, Hunan, People's Republic of China
| | - Ji-Ye Yin
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha 410078, People's Republic of China.,Institute of Clinical Pharmacology, Central South University; Hunan Key Laboratory of Pharmacogenetics, Changsha 410078, People's Republic of China.,Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha 410078, People's Republic of China.,Department of Geratic Surgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, People's Republic of China
| | - Wei Wu
- Department of Geratic Surgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, People's Republic of China.,National Clinical Research Center for Geriatric Disorders, Changsha, Hunan 410008, People's Republic of China
| |
Collapse
|
12
|
Zhao X, Mu C, Ma J, Dai X, Jiao H. The association of four SNPs in DNA mismatch repair genes with idiopathic male infertility in northwest China. Int J Immunogenet 2019; 46:451-458. [DOI: 10.1111/iji.12448] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Revised: 05/23/2019] [Accepted: 06/10/2019] [Indexed: 12/14/2022]
Affiliation(s)
- Xinyan Zhao
- Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education Ningxia Medical University Yinchuan China
- Key Laboratory of Reproduction and Genetics in Ningxia Yinchuan China
- Department of Medical Genetic and Cell Biology, Basic Medicine Science College Ningxia Medical University Yinchuan China
| | - Chunlan Mu
- Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education Ningxia Medical University Yinchuan China
- Key Laboratory of Reproduction and Genetics in Ningxia Yinchuan China
- Department of Medical Genetic and Cell Biology, Basic Medicine Science College Ningxia Medical University Yinchuan China
| | - Jia Ma
- Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education Ningxia Medical University Yinchuan China
- Key Laboratory of Reproduction and Genetics in Ningxia Yinchuan China
- Department of Medical Genetic and Cell Biology, Basic Medicine Science College Ningxia Medical University Yinchuan China
| | - Xiaojing Dai
- Health Science Center University of Texas Houston Texas USA
| | - Haiyan Jiao
- Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education Ningxia Medical University Yinchuan China
- Key Laboratory of Reproduction and Genetics in Ningxia Yinchuan China
- Department of Medical Genetic and Cell Biology, Basic Medicine Science College Ningxia Medical University Yinchuan China
| |
Collapse
|
13
|
Zhang X, Ding K, Wang J, Li X, Zhao P. Chemoresistance caused by the microenvironment of glioblastoma and the corresponding solutions. Biomed Pharmacother 2018; 109:39-46. [PMID: 30391707 DOI: 10.1016/j.biopha.2018.10.063] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Revised: 10/03/2018] [Accepted: 10/12/2018] [Indexed: 12/30/2022] Open
Abstract
Glioblastoma multiforme (GBM) is the most common and aggressive primary human brain tumor. Although comprehensive therapies combining radiotherapy and chemotherapy after surgery can prolong survival, the prognosis is still poor with a median survival of only 14.6 months. Chemoresistance is one of the major causes of relapse as well as poor survival in glioma patients. Therefore, novel strategies to overcome chemoresistance are desperately needed for improved treatment of human GBM. Recent studies have demonstrated that the tumor microenvironment plays a critical role in the chemoresistance of various tumor types, which makes it a suitable target in anti-cancer therapies, as well as a valuable biomarker for prognostic purposes. This review focuses on chemoresistance in GBM induced by stromal cells, including the endothelium of blood vessels, astrocytes, and myeloid cells, as well as non-cellular factors in the tumor microenvironment. Corresponding therapies are discussed, including progressive strategies involving 3-dimensional models integrating engineering as well as biological advances.
Collapse
Affiliation(s)
- Xin Zhang
- Department of Neurosurgery, Qilu Hospital of Shandong University, Institute of Brain and Brain-Inspired Science, Shandong University, PR China; Shandong Key Laboratory of Brain Function Remodeling, PR China
| | - Kaikai Ding
- Shandong Key Laboratory of Brain Function Remodeling, PR China; Department of Radiation Oncology, Qilu Hospital of Shandong University, Jinan, 250012, PR China
| | - Jian Wang
- Department of Neurosurgery, Qilu Hospital of Shandong University, Institute of Brain and Brain-Inspired Science, Shandong University, PR China; Shandong Key Laboratory of Brain Function Remodeling, PR China; Department of Biomedicine, University of Bergen, 5009, Bergen, Norway
| | - Xingang Li
- Department of Neurosurgery, Qilu Hospital of Shandong University, Institute of Brain and Brain-Inspired Science, Shandong University, PR China; Shandong Key Laboratory of Brain Function Remodeling, PR China
| | - Peng Zhao
- Department of Neurosurgery, Qilu Hospital of Shandong University, Institute of Brain and Brain-Inspired Science, Shandong University, PR China; Shandong Key Laboratory of Brain Function Remodeling, PR China.
| |
Collapse
|
14
|
Kaur K, Kaur R. Absence of APE1 (Asp148Glu) gene polymorphism in North-West Indian population: A comparison with world population. Meta Gene 2018. [DOI: 10.1016/j.mgene.2018.03.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022] Open
|
15
|
Qian X, Tan H, Zhang J, Liu K, Yang T, Wang M, Debinskie W, Zhao W, Chan MD, Zhou X. Identification of biomarkers for pseudo and true progression of GBM based on radiogenomics study. Oncotarget 2018; 7:55377-55394. [PMID: 27421136 PMCID: PMC5342424 DOI: 10.18632/oncotarget.10553] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Accepted: 05/05/2016] [Indexed: 02/06/2023] Open
Abstract
The diagnosis for pseudoprogression (PsP) and true tumor progression (TTP) of GBM is a challenging task in clinical practices. The purpose of this study is to identify potential genetic biomarkers associated with PsP and TTP based on the clinical records, longitudinal imaging features, and genomics data. We are the first to introduce the radiogenomics approach to identify candidate genes for PsP and TTP of GBM. Specifically, a novel longitudinal sparse regression model was developed to construct the relationship between gene expression and imaging features. The imaging features were extracted from tumors along the longitudinal MRI and provided diagnostic information of PsP and TTP. The 33 candidate genes were selected based on their association with the imaging features, reflecting their relation with the development of PsP and TTP. We then conducted biological relevance analysis for 33 candidate genes to identify the potential biomarkers, i.e., Interferon regulatory factor (IRF9) and X-ray repair cross-complementing gene (XRCC1), which were involved in the cancer suppression and prevention, respectively. The IRF9 and XRCC1 were further independently validated in the TCGA data. Our results provided the first substantial evidence that IRF9 and XRCC1 can serve as the potential biomarkers for the development of PsP and TTP.
Collapse
Affiliation(s)
- Xiaohua Qian
- Department of Radiology, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
| | - Hua Tan
- Department of Radiology, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
| | - Jian Zhang
- Department of Radiology, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
| | - Keqin Liu
- Department of Radiology, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
| | - Tielin Yang
- School of Life Science, Xi'an Jiaotong University, Xi'an, Shanxi 710049, China
| | - Maode Wang
- The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shanxi 710061, China
| | - Waldemar Debinskie
- Department of Radiation Oncology, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
| | - Weilin Zhao
- Department of Radiology, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
| | - Michael D Chan
- Department of Radiation Oncology, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
| | - Xiaobo Zhou
- Department of Radiology, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
| |
Collapse
|
16
|
Li S, Zheng Y, Tian T, Wang M, Liu X, Liu K, Zhai Y, Dai C, Deng Y, Li S, Dai Z, Lu J. Pooling-analysis on hMLH1 polymorphisms and cancer risk: evidence based on 31,484 cancer cases and 45,494 cancer-free controls. Oncotarget 2017; 8:93063-93078. [PMID: 29190978 PMCID: PMC5696244 DOI: 10.18632/oncotarget.21810] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Accepted: 09/08/2017] [Indexed: 01/18/2023] Open
Abstract
To elucidate the veritable relationship between three hMLH1 polymorphisms (rs1800734, rs1799977, rs63750447) and cancer risk, we performed this meta-analysis based on overall published data up to May 2017, from PubMed, Web of knowledge, VIP, WanFang and CNKI database, and the references of the original studies or review articles. 57 publications including 31,484 cancer cases and 45,494 cancer-free controls were obtained. The quality assessment of six articles obtained a summarized score less than 6 in terms of the Newcastle-Ottawa Scale (NOS). All statistical analyses were calculated with the software STATA (Version 14.0; Stata Corp, College Station, TX). We found all the three polymorphisms can enhance overall cancer risk, especially in Asians, under different genetic comparisons. In the subgroup analysis by cancer type, we found a moderate association between rs1800734 and the risk of gastric cancer (allele model: OR = 1.14, P = 0.017; homozygote model: OR = 1.33, P = 0.019; dominant model: OR = 1.27, P = 0.024) and lung cancer in recessive model (OR = 1.27, P = 0.024). The G allele of rs1799977 polymorphism was proved to connect with susceptibility of colorectal cancer (allele model: OR = 1.21, P = 0.023; dominate model: OR = 1.32, P <0.0001) and prostate cancer (dominate model: OR = 1.36, P <0.0001). Rs63750447 showed an increased risk of colorectal cancer, endometrial cancer and gastric cancer under all genetic models. These findings provide evidence that hMLH1 polymorphisms may associate with cancer risk, especially in Asians.
Collapse
Affiliation(s)
- Sha Li
- Clinical Research Center, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China.,Department of Pharmacy, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China
| | - Yi Zheng
- Clinical Research Center, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China.,Department of Oncology, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China
| | - Tian Tian
- Department of Oncology, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China
| | - Meng Wang
- Department of Oncology, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China
| | - Xinghan Liu
- Department of Oncology, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China
| | - Kang Liu
- Department of Oncology, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China
| | - Yajing Zhai
- Clinical Research Center, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Cong Dai
- Department of Oncology, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China
| | - Yujiao Deng
- Department of Oncology, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China
| | - Shanli Li
- Department of Oncology, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China
| | - Zhijun Dai
- Department of Oncology, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China
| | - Jun Lu
- Clinical Research Center, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| |
Collapse
|
17
|
Wang X, Sun CL, Hageman L, Smith K, Singh P, Desai S, Hawkins DS, Hudson MM, Mascarenhas L, Neglia JP, Oeffinger KC, Ritchey AK, Robison LL, Villaluna D, Landier W, Bhatia S. Clinical and Genetic Risk Prediction of Subsequent CNS Tumors in Survivors of Childhood Cancer: A Report From the COG ALTE03N1 Study. J Clin Oncol 2017; 35:3688-3696. [PMID: 28976792 DOI: 10.1200/jco.2017.74.7444] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Purpose Survivors of childhood cancer treated with cranial radiation therapy are at risk for subsequent CNS tumors. However, significant interindividual variability in risk suggests a role for genetic susceptibility and provides an opportunity to identify survivors of childhood cancer at increased risk for these tumors. Methods We curated candidate genetic variants from previously published studies in adult-onset primary CNS tumors and replicated these in survivors of childhood cancer with and without subsequent CNS tumors (82 participants and 228 matched controls). We developed prediction models to identify survivors at high or low risk for subsequent CNS tumors and validated these models in an independent matched case-control sample (25 participants and 54 controls). Results We demonstrated an association between six previously published single nucleotide polymorphisms (rs15869 [ BRCA2], rs1805389 [ LIG4], rs8079544 [ TP53], rs25489 [ XRCC1], rs1673041 [ POLD1], and rs11615 [ ERCC1]) and subsequent CNS tumors in survivors of childhood cancer. Including genetic variants in a Final Model containing age at primary cancer, sex, and cranial radiation therapy dose yielded an area under the curve of 0.81 (95% CI, 0.76 to 0.86), which was superior ( P = .002) to the Clinical Model (area under the curve, 0.73; 95% CI, 0.66 to 0.80). The prediction model was successfully validated. The sensitivity and specificity of predicting survivors of childhood cancer at highest or lowest risk of subsequent CNS tumors was 87.5% and 83.5%, respectively. Conclusion It is possible to identify survivors of childhood cancer at high or low risk for subsequent CNS tumors on the basis of genetic and clinical information. This information can be used to inform surveillance for early detection of subsequent CNS tumors.
Collapse
Affiliation(s)
- Xuexia Wang
- Xuexia Wang, University of North Texas, Denton, TX; Can-Lan Sun, City of Hope, Duarte; Leo Mascarenhas, Children's Hospital Los Angeles, University of Southern California, Los Angeles; Doojduen Villaluna, Children's Oncology Group, Monrovia, CA; Lindsey Hageman, Kandice Smith, Purnima Singh, Wendy Landier, and Smita Bhatia, University of Alabama at Birmingham, Birmingham, AL; Sunil Desai, University of Alberta, Edmonton, Alberta, Canada; Douglas S. Hawkins, Seattle Children's Hospital, University of Washington, Fred Hutchinson Cancer Research Center, Seattle, WA; Melissa M. Hudson and Leslie L. Robison, St Jude Children's Research Hospital, Memphis, TN; Joseph P. Neglia, University of Minnesota Medical School, Minneapolis, MN; Kevin C. Oeffinger, Duke University Medical Center, Durham, NC; and A. Kim Ritchey, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Can-Lan Sun
- Xuexia Wang, University of North Texas, Denton, TX; Can-Lan Sun, City of Hope, Duarte; Leo Mascarenhas, Children's Hospital Los Angeles, University of Southern California, Los Angeles; Doojduen Villaluna, Children's Oncology Group, Monrovia, CA; Lindsey Hageman, Kandice Smith, Purnima Singh, Wendy Landier, and Smita Bhatia, University of Alabama at Birmingham, Birmingham, AL; Sunil Desai, University of Alberta, Edmonton, Alberta, Canada; Douglas S. Hawkins, Seattle Children's Hospital, University of Washington, Fred Hutchinson Cancer Research Center, Seattle, WA; Melissa M. Hudson and Leslie L. Robison, St Jude Children's Research Hospital, Memphis, TN; Joseph P. Neglia, University of Minnesota Medical School, Minneapolis, MN; Kevin C. Oeffinger, Duke University Medical Center, Durham, NC; and A. Kim Ritchey, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Lindsey Hageman
- Xuexia Wang, University of North Texas, Denton, TX; Can-Lan Sun, City of Hope, Duarte; Leo Mascarenhas, Children's Hospital Los Angeles, University of Southern California, Los Angeles; Doojduen Villaluna, Children's Oncology Group, Monrovia, CA; Lindsey Hageman, Kandice Smith, Purnima Singh, Wendy Landier, and Smita Bhatia, University of Alabama at Birmingham, Birmingham, AL; Sunil Desai, University of Alberta, Edmonton, Alberta, Canada; Douglas S. Hawkins, Seattle Children's Hospital, University of Washington, Fred Hutchinson Cancer Research Center, Seattle, WA; Melissa M. Hudson and Leslie L. Robison, St Jude Children's Research Hospital, Memphis, TN; Joseph P. Neglia, University of Minnesota Medical School, Minneapolis, MN; Kevin C. Oeffinger, Duke University Medical Center, Durham, NC; and A. Kim Ritchey, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Kandice Smith
- Xuexia Wang, University of North Texas, Denton, TX; Can-Lan Sun, City of Hope, Duarte; Leo Mascarenhas, Children's Hospital Los Angeles, University of Southern California, Los Angeles; Doojduen Villaluna, Children's Oncology Group, Monrovia, CA; Lindsey Hageman, Kandice Smith, Purnima Singh, Wendy Landier, and Smita Bhatia, University of Alabama at Birmingham, Birmingham, AL; Sunil Desai, University of Alberta, Edmonton, Alberta, Canada; Douglas S. Hawkins, Seattle Children's Hospital, University of Washington, Fred Hutchinson Cancer Research Center, Seattle, WA; Melissa M. Hudson and Leslie L. Robison, St Jude Children's Research Hospital, Memphis, TN; Joseph P. Neglia, University of Minnesota Medical School, Minneapolis, MN; Kevin C. Oeffinger, Duke University Medical Center, Durham, NC; and A. Kim Ritchey, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Purnima Singh
- Xuexia Wang, University of North Texas, Denton, TX; Can-Lan Sun, City of Hope, Duarte; Leo Mascarenhas, Children's Hospital Los Angeles, University of Southern California, Los Angeles; Doojduen Villaluna, Children's Oncology Group, Monrovia, CA; Lindsey Hageman, Kandice Smith, Purnima Singh, Wendy Landier, and Smita Bhatia, University of Alabama at Birmingham, Birmingham, AL; Sunil Desai, University of Alberta, Edmonton, Alberta, Canada; Douglas S. Hawkins, Seattle Children's Hospital, University of Washington, Fred Hutchinson Cancer Research Center, Seattle, WA; Melissa M. Hudson and Leslie L. Robison, St Jude Children's Research Hospital, Memphis, TN; Joseph P. Neglia, University of Minnesota Medical School, Minneapolis, MN; Kevin C. Oeffinger, Duke University Medical Center, Durham, NC; and A. Kim Ritchey, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Sunil Desai
- Xuexia Wang, University of North Texas, Denton, TX; Can-Lan Sun, City of Hope, Duarte; Leo Mascarenhas, Children's Hospital Los Angeles, University of Southern California, Los Angeles; Doojduen Villaluna, Children's Oncology Group, Monrovia, CA; Lindsey Hageman, Kandice Smith, Purnima Singh, Wendy Landier, and Smita Bhatia, University of Alabama at Birmingham, Birmingham, AL; Sunil Desai, University of Alberta, Edmonton, Alberta, Canada; Douglas S. Hawkins, Seattle Children's Hospital, University of Washington, Fred Hutchinson Cancer Research Center, Seattle, WA; Melissa M. Hudson and Leslie L. Robison, St Jude Children's Research Hospital, Memphis, TN; Joseph P. Neglia, University of Minnesota Medical School, Minneapolis, MN; Kevin C. Oeffinger, Duke University Medical Center, Durham, NC; and A. Kim Ritchey, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Douglas S Hawkins
- Xuexia Wang, University of North Texas, Denton, TX; Can-Lan Sun, City of Hope, Duarte; Leo Mascarenhas, Children's Hospital Los Angeles, University of Southern California, Los Angeles; Doojduen Villaluna, Children's Oncology Group, Monrovia, CA; Lindsey Hageman, Kandice Smith, Purnima Singh, Wendy Landier, and Smita Bhatia, University of Alabama at Birmingham, Birmingham, AL; Sunil Desai, University of Alberta, Edmonton, Alberta, Canada; Douglas S. Hawkins, Seattle Children's Hospital, University of Washington, Fred Hutchinson Cancer Research Center, Seattle, WA; Melissa M. Hudson and Leslie L. Robison, St Jude Children's Research Hospital, Memphis, TN; Joseph P. Neglia, University of Minnesota Medical School, Minneapolis, MN; Kevin C. Oeffinger, Duke University Medical Center, Durham, NC; and A. Kim Ritchey, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Melissa M Hudson
- Xuexia Wang, University of North Texas, Denton, TX; Can-Lan Sun, City of Hope, Duarte; Leo Mascarenhas, Children's Hospital Los Angeles, University of Southern California, Los Angeles; Doojduen Villaluna, Children's Oncology Group, Monrovia, CA; Lindsey Hageman, Kandice Smith, Purnima Singh, Wendy Landier, and Smita Bhatia, University of Alabama at Birmingham, Birmingham, AL; Sunil Desai, University of Alberta, Edmonton, Alberta, Canada; Douglas S. Hawkins, Seattle Children's Hospital, University of Washington, Fred Hutchinson Cancer Research Center, Seattle, WA; Melissa M. Hudson and Leslie L. Robison, St Jude Children's Research Hospital, Memphis, TN; Joseph P. Neglia, University of Minnesota Medical School, Minneapolis, MN; Kevin C. Oeffinger, Duke University Medical Center, Durham, NC; and A. Kim Ritchey, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Leo Mascarenhas
- Xuexia Wang, University of North Texas, Denton, TX; Can-Lan Sun, City of Hope, Duarte; Leo Mascarenhas, Children's Hospital Los Angeles, University of Southern California, Los Angeles; Doojduen Villaluna, Children's Oncology Group, Monrovia, CA; Lindsey Hageman, Kandice Smith, Purnima Singh, Wendy Landier, and Smita Bhatia, University of Alabama at Birmingham, Birmingham, AL; Sunil Desai, University of Alberta, Edmonton, Alberta, Canada; Douglas S. Hawkins, Seattle Children's Hospital, University of Washington, Fred Hutchinson Cancer Research Center, Seattle, WA; Melissa M. Hudson and Leslie L. Robison, St Jude Children's Research Hospital, Memphis, TN; Joseph P. Neglia, University of Minnesota Medical School, Minneapolis, MN; Kevin C. Oeffinger, Duke University Medical Center, Durham, NC; and A. Kim Ritchey, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Joseph P Neglia
- Xuexia Wang, University of North Texas, Denton, TX; Can-Lan Sun, City of Hope, Duarte; Leo Mascarenhas, Children's Hospital Los Angeles, University of Southern California, Los Angeles; Doojduen Villaluna, Children's Oncology Group, Monrovia, CA; Lindsey Hageman, Kandice Smith, Purnima Singh, Wendy Landier, and Smita Bhatia, University of Alabama at Birmingham, Birmingham, AL; Sunil Desai, University of Alberta, Edmonton, Alberta, Canada; Douglas S. Hawkins, Seattle Children's Hospital, University of Washington, Fred Hutchinson Cancer Research Center, Seattle, WA; Melissa M. Hudson and Leslie L. Robison, St Jude Children's Research Hospital, Memphis, TN; Joseph P. Neglia, University of Minnesota Medical School, Minneapolis, MN; Kevin C. Oeffinger, Duke University Medical Center, Durham, NC; and A. Kim Ritchey, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Kevin C Oeffinger
- Xuexia Wang, University of North Texas, Denton, TX; Can-Lan Sun, City of Hope, Duarte; Leo Mascarenhas, Children's Hospital Los Angeles, University of Southern California, Los Angeles; Doojduen Villaluna, Children's Oncology Group, Monrovia, CA; Lindsey Hageman, Kandice Smith, Purnima Singh, Wendy Landier, and Smita Bhatia, University of Alabama at Birmingham, Birmingham, AL; Sunil Desai, University of Alberta, Edmonton, Alberta, Canada; Douglas S. Hawkins, Seattle Children's Hospital, University of Washington, Fred Hutchinson Cancer Research Center, Seattle, WA; Melissa M. Hudson and Leslie L. Robison, St Jude Children's Research Hospital, Memphis, TN; Joseph P. Neglia, University of Minnesota Medical School, Minneapolis, MN; Kevin C. Oeffinger, Duke University Medical Center, Durham, NC; and A. Kim Ritchey, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - A Kim Ritchey
- Xuexia Wang, University of North Texas, Denton, TX; Can-Lan Sun, City of Hope, Duarte; Leo Mascarenhas, Children's Hospital Los Angeles, University of Southern California, Los Angeles; Doojduen Villaluna, Children's Oncology Group, Monrovia, CA; Lindsey Hageman, Kandice Smith, Purnima Singh, Wendy Landier, and Smita Bhatia, University of Alabama at Birmingham, Birmingham, AL; Sunil Desai, University of Alberta, Edmonton, Alberta, Canada; Douglas S. Hawkins, Seattle Children's Hospital, University of Washington, Fred Hutchinson Cancer Research Center, Seattle, WA; Melissa M. Hudson and Leslie L. Robison, St Jude Children's Research Hospital, Memphis, TN; Joseph P. Neglia, University of Minnesota Medical School, Minneapolis, MN; Kevin C. Oeffinger, Duke University Medical Center, Durham, NC; and A. Kim Ritchey, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Leslie L Robison
- Xuexia Wang, University of North Texas, Denton, TX; Can-Lan Sun, City of Hope, Duarte; Leo Mascarenhas, Children's Hospital Los Angeles, University of Southern California, Los Angeles; Doojduen Villaluna, Children's Oncology Group, Monrovia, CA; Lindsey Hageman, Kandice Smith, Purnima Singh, Wendy Landier, and Smita Bhatia, University of Alabama at Birmingham, Birmingham, AL; Sunil Desai, University of Alberta, Edmonton, Alberta, Canada; Douglas S. Hawkins, Seattle Children's Hospital, University of Washington, Fred Hutchinson Cancer Research Center, Seattle, WA; Melissa M. Hudson and Leslie L. Robison, St Jude Children's Research Hospital, Memphis, TN; Joseph P. Neglia, University of Minnesota Medical School, Minneapolis, MN; Kevin C. Oeffinger, Duke University Medical Center, Durham, NC; and A. Kim Ritchey, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Doojduen Villaluna
- Xuexia Wang, University of North Texas, Denton, TX; Can-Lan Sun, City of Hope, Duarte; Leo Mascarenhas, Children's Hospital Los Angeles, University of Southern California, Los Angeles; Doojduen Villaluna, Children's Oncology Group, Monrovia, CA; Lindsey Hageman, Kandice Smith, Purnima Singh, Wendy Landier, and Smita Bhatia, University of Alabama at Birmingham, Birmingham, AL; Sunil Desai, University of Alberta, Edmonton, Alberta, Canada; Douglas S. Hawkins, Seattle Children's Hospital, University of Washington, Fred Hutchinson Cancer Research Center, Seattle, WA; Melissa M. Hudson and Leslie L. Robison, St Jude Children's Research Hospital, Memphis, TN; Joseph P. Neglia, University of Minnesota Medical School, Minneapolis, MN; Kevin C. Oeffinger, Duke University Medical Center, Durham, NC; and A. Kim Ritchey, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Wendy Landier
- Xuexia Wang, University of North Texas, Denton, TX; Can-Lan Sun, City of Hope, Duarte; Leo Mascarenhas, Children's Hospital Los Angeles, University of Southern California, Los Angeles; Doojduen Villaluna, Children's Oncology Group, Monrovia, CA; Lindsey Hageman, Kandice Smith, Purnima Singh, Wendy Landier, and Smita Bhatia, University of Alabama at Birmingham, Birmingham, AL; Sunil Desai, University of Alberta, Edmonton, Alberta, Canada; Douglas S. Hawkins, Seattle Children's Hospital, University of Washington, Fred Hutchinson Cancer Research Center, Seattle, WA; Melissa M. Hudson and Leslie L. Robison, St Jude Children's Research Hospital, Memphis, TN; Joseph P. Neglia, University of Minnesota Medical School, Minneapolis, MN; Kevin C. Oeffinger, Duke University Medical Center, Durham, NC; and A. Kim Ritchey, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Smita Bhatia
- Xuexia Wang, University of North Texas, Denton, TX; Can-Lan Sun, City of Hope, Duarte; Leo Mascarenhas, Children's Hospital Los Angeles, University of Southern California, Los Angeles; Doojduen Villaluna, Children's Oncology Group, Monrovia, CA; Lindsey Hageman, Kandice Smith, Purnima Singh, Wendy Landier, and Smita Bhatia, University of Alabama at Birmingham, Birmingham, AL; Sunil Desai, University of Alberta, Edmonton, Alberta, Canada; Douglas S. Hawkins, Seattle Children's Hospital, University of Washington, Fred Hutchinson Cancer Research Center, Seattle, WA; Melissa M. Hudson and Leslie L. Robison, St Jude Children's Research Hospital, Memphis, TN; Joseph P. Neglia, University of Minnesota Medical School, Minneapolis, MN; Kevin C. Oeffinger, Duke University Medical Center, Durham, NC; and A. Kim Ritchey, University of Pittsburgh School of Medicine, Pittsburgh, PA
| |
Collapse
|
18
|
Qian T, Zhang B, Qian C, He Y, Li Y. Association between common polymorphisms in ERCC gene and glioma risk: A meta-analysis of 15 studies. Medicine (Baltimore) 2017; 96:e6832. [PMID: 28514298 PMCID: PMC5440135 DOI: 10.1097/md.0000000000006832] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND A number of studies have investigated the roles of excision repair cross complementation group 1 (ERCC1), ERCC2, and ERCC5 genes polymorphisms in the development of glioma; however, the results were inconsistent. Here, we performed a meta-analysis to investigate the association between 6 polymorphisms in the ERCC genes (rs3212986, rs11615, rs13181, rs1799793, rs238406, rs17655) and glioma risk. METHODS The PubMed, Embase, and Web of science were searched up to September 6, 2016, for studies on the association between ERCC polymorphisms and glioma risk. A fixed-effects or random-effects model was used to calculate the pooled odds ratios based on the results from the heterogeneity tests. Sensitivity and cumulative meta-analyses were also performed. RESULTS A total of 15 studies were eligible for the pooled analysis, conducted in 2 populations of ethnic descent: 8 Europeans and 7 Asians. The results showed that ERCC1 rs3212986 polymorphism was positively associated with glioma [AA vs CC: odds ratio (OR) = 1.298, 95% confidence interval (95% CI) = 1.043-1.230, P = .025]. Association of the ERCC2 rs13181 and rs1799793 polymorphisms was only observed in Asians (CC vs AA for rs13181: OR = 1.539, 95% CI = 1.122-2.109, P = .007; AA vs GG for rs1799793: OR = 1.474, 95% CI = 1.090-1.994, P = .012). However, no association was observed between glioma risk and ERCC1 rs11615, ERCC2 rs238406, and ERCC5 rs17655 polymorphisms. Moreover, sensitivity and cumulative meta-analyses confirmed the stability of the results. CONCLUSIONS Our meta-analysis indicated that the ERCC1 rs3212986 polymorphism and 2 polymorphisms in ERCC2 gene (rs13181 and rs1799793) contributed to the susceptibility of glioma.
Collapse
|
19
|
Savio AJ, Bapat B. Modulation of transcription factor binding and epigenetic regulation of the MLH1 CpG island and shore by polymorphism rs1800734 in colorectal cancer. Epigenetics 2017; 12:441-448. [PMID: 28304185 DOI: 10.1080/15592294.2017.1305527] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The MLH1 promoter polymorphism rs1800734 is associated with MLH1 CpG island hypermethylation and expression loss in colorectal cancer (CRC). Conversely, variant rs1800734 is associated with MLH1 shore, but not island, hypomethylation in peripheral blood mononuclear cell DNA. To explore these distinct patterns, MLH1 CpG island and shore methylation was assessed in CRC cell lines stratified by rs1800734 genotype. Cell lines containing the variant A allele demonstrated MLH1 shore hypomethylation compared to wild type (GG). There was significant enrichment of transcription factor AP4 at the MLH1 promoter in GG and GA cell lines, but not the AA cell line, by chromatin immunoprecipitation studies. Preferential binding to the G allele was confirmed by sequencing in the GA cell line. The enhancer-associated histone modification H3K4me1 was enriched at the MLH1 shore; however, H3K27ac was not, indicating the shore is an inactive enhancer. These results demonstrate the role of variant rs1800734 in altering transcription factor binding as well as epigenetics at regions beyond the MLH1 CpG island in which it is located.
Collapse
Affiliation(s)
- Andrea J Savio
- a Lunenfeld-Tanenbaum Research Institute, Sinai Health System , Toronto , Ontario , Canada.,b Department of Laboratory Medicine and Pathobiology , University of Toronto , Toronto , Ontario , Canada
| | - Bharati Bapat
- a Lunenfeld-Tanenbaum Research Institute, Sinai Health System , Toronto , Ontario , Canada.,b Department of Laboratory Medicine and Pathobiology , University of Toronto , Toronto , Ontario , Canada.,c Department of Pathology , University Health Network , Toronto , Ontario , Canada
| |
Collapse
|
20
|
Liu K, Jiang Y. Polymorphisms in DNA Repair Gene and Susceptibility to Glioma: A Systematic Review and Meta-Analysis Based on 33 Studies with 15 SNPs in 9 Genes. Cell Mol Neurobiol 2017; 37:263-274. [PMID: 27055523 DOI: 10.1007/s10571-016-0367-y] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2016] [Accepted: 03/22/2016] [Indexed: 11/29/2022]
Abstract
At present, many publications have evaluated the correlation between the DNA repair gene polymorphisms and glioma susceptibility. However, the results remain inconclusive. The aim of this research is to exhaustively assess the association of genetic polymorphisms in DNA repair genes with glioma risk in human. Meta-analysis method was conducted, and 33 studies with 15 SNPs in 9 genes were included (12553 glioma cases and 17178 controls). Correlation strength was evaluated by odds ratio with a 95 % confidence interval. Rs1799782 T allele and rs25487A allele might bring about higher risk of glioma in Asian population. Rs1805377 G allele was an increased risk genetic factor of glioma. Asian carried with rs3212986 A allele was more likely to have glioma. Rs1800067 G allele was a risk factor of developing glioma. Carriers with rs12917 CC genotype in MGMT gene had higher risk of glioma in Caucasian than other non-CC genotype carriers. Carriers with rs1136410 T allele in PARP1 gene could more likely to develop glioma in Caucasian. This meta-analysis suggests that glioma susceptibility is associated with rs1799782 and rs25487 of X-ray repair complementing defective repair in Chinese hamster cells 1 (XRCC1), rs1805377 of XRCC4, rs1800067 of excision repair cross-complementing rodent repair deficiency complementation group 4 (ERCC4) and rs3212986 of ERCC1 in Asian population, and rs12917 of O-6-methylguanine-DNA methyltransferase (MGMT) and rs1136410 of poly(ADP-ribose) polymerase 1 (PARP1) in Caucasian population.
Collapse
Affiliation(s)
- Kun Liu
- Department of neurosurgery, The Second Xiangya Hospital of Central South University, 139 Renmin(M) Road, Changsha, 410011, Hunan, China
- Department of neurosurgery, Brains Hospital of Hunan Province, Changsha, China
| | - Yugang Jiang
- Department of neurosurgery, The Second Xiangya Hospital of Central South University, 139 Renmin(M) Road, Changsha, 410011, Hunan, China.
| |
Collapse
|
21
|
Liu NQ, ter Huurne M, Nguyen LN, Peng T, Wang SY, Studd JB, Joshi O, Ongen H, Bramsen JB, Yan J, Andersen CL, Taipale J, Dermitzakis ET, Houlston RS, Hubner NC, Stunnenberg HG. The non-coding variant rs1800734 enhances DCLK3 expression through long-range interaction and promotes colorectal cancer progression. Nat Commun 2017; 8:14418. [PMID: 28195176 PMCID: PMC5316867 DOI: 10.1038/ncomms14418] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Accepted: 12/28/2016] [Indexed: 01/02/2023] Open
Abstract
Genome-wide association studies have identified a great number of non-coding risk variants for colorectal cancer (CRC). To date, the majority of these variants have not been functionally studied. Identification of allele-specific transcription factor (TF) binding is of great importance to understand regulatory consequences of such variants. A recently developed proteome-wide analysis of disease-associated SNPs (PWAS) enables identification of TF-DNA interactions in an unbiased manner. Here we perform a large-scale PWAS study to comprehensively characterize TF-binding landscape that is associated with CRC, which identifies 731 allele-specific TF binding at 116 CRC risk loci. This screen identifies the A-allele of rs1800734 within the promoter region of MLH1 as perturbing the binding of TFAP4 and consequently increasing DCLK3 expression through a long-range interaction, which promotes cancer malignancy through enhancing expression of the genes related to epithelial-to-mesenchymal transition.
Collapse
Affiliation(s)
- Ning Qing Liu
- Faculty of Science, Department of Molecular Biology, Radboud University, RIMLS, PO BOX 9101, 6500HB Nijmegen, The Netherlands
| | - Menno ter Huurne
- Faculty of Science, Department of Molecular Biology, Radboud University, RIMLS, PO BOX 9101, 6500HB Nijmegen, The Netherlands
| | - Luan N. Nguyen
- Faculty of Science, Department of Molecular Biology, Radboud University, RIMLS, PO BOX 9101, 6500HB Nijmegen, The Netherlands
| | - Tianran Peng
- Faculty of Science, Department of Molecular Biology, Radboud University, RIMLS, PO BOX 9101, 6500HB Nijmegen, The Netherlands
| | - Shuang-Yin Wang
- Faculty of Science, Department of Molecular Biology, Radboud University, RIMLS, PO BOX 9101, 6500HB Nijmegen, The Netherlands
| | - James B. Studd
- Division of Genetics and Epidemiology, Institute of Cancer Research, 15 Cotswold Road, Sutton, SM2 5NG Surrey, UK
| | - Onkar Joshi
- Faculty of Science, Department of Molecular Biology, Radboud University, RIMLS, PO BOX 9101, 6500HB Nijmegen, The Netherlands
| | - Halit Ongen
- Department of Genetic Medicine and Development, University of Geneva Medical School, Geneva 1211, Switzerland
| | - Jesper B Bramsen
- Department of Molecular Medicine, Aarhus University Hospital, Palle Juul-Jensens Boulevard 99, DK-8200 Aarhus, Denmark
| | - Jian Yan
- Division of Functional Genomics and Systems Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, SE 141 83 Stockholm, Sweden
- Ludwig Institute for Cancer Research, 9500 Gilman Drive, La Jolla, California 92093, USA
| | - Claus L. Andersen
- Department of Molecular Medicine, Aarhus University Hospital, Palle Juul-Jensens Boulevard 99, DK-8200 Aarhus, Denmark
| | - Jussi Taipale
- Division of Functional Genomics and Systems Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, SE 141 83 Stockholm, Sweden
| | - Emmanouil T. Dermitzakis
- Department of Genetic Medicine and Development, University of Geneva Medical School, Geneva 1211, Switzerland
| | - Richard S. Houlston
- Division of Genetics and Epidemiology, Institute of Cancer Research, 15 Cotswold Road, Sutton, SM2 5NG Surrey, UK
| | - Nina C. Hubner
- Faculty of Science, Department of Molecular Biology, Radboud University, RIMLS, PO BOX 9101, 6500HB Nijmegen, The Netherlands
| | - Hendrik G. Stunnenberg
- Faculty of Science, Department of Molecular Biology, Radboud University, RIMLS, PO BOX 9101, 6500HB Nijmegen, The Netherlands
| |
Collapse
|
22
|
Lee SY, Hong MJ, Jeon HS, Choi YY, Choi JE, Kang HG, Jung DK, Jin C, Do SK, Yoo SS, Seok Y, Lee EB, Shin KM, Jeong JY, Lee WK, Lee J, Cha SI, Kim CH, Kim YT, Jheon S, Park JY. Functional intronic ERCC1 polymorphism from regulomeDB can predict survival in lung cancer after surgery. Oncotarget 2016; 6:24522-32. [PMID: 26056042 PMCID: PMC4695203 DOI: 10.18632/oncotarget.4083] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2015] [Accepted: 05/13/2015] [Indexed: 11/25/2022] Open
Abstract
We searched for potential regulatory single nucleotide polymorphisms (SNPs) in excision repair cross-complementing group 1 (ERCC1) using RegulomeDB, a database integrating information from the Encyclopedia of DNA Elements (ENCODE) project, and investigated their association with survival after surgery in non-small cell lung cancer (NSCLC). Among 364 SNPs found within ERCC1 region using RegulomeDB, four top priority SNPs (rs2298881C>A, rs1049739A>G, rs10415949A>G and rs6509214G>T) were selected for this study. The four SNPs were investigated in 316 patients. A replication study was performed (n = 579). Of the four SNPs analyzed in the discovery set, rs2298881C>A and rs6509214G>T were significantly associated with survival outcomes. The association was consistently observed only for rs2298881C>A in the validation cohort. In combined analysis, rs2298881C>A was significantly associated with worse overall survival and disease-free survival (P = 0.0002 and 0.02, respectively). A decreased reporter gene expression for rs2298881 A allele was observed compared with C allele by luciferase assay (P = 0.02). ERCC1 rs2298881C>A, an intronic SNP, is the first genetic polymorphism with functional evidence of regulating its expression, and the SNP is associated with prognosis of NSCLC. Our result supports the role of RegulomeDB as a comprehensive source of prioritized candidate SNPs for genetic association studies.
Collapse
Affiliation(s)
- Shin Yup Lee
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Daegu, Republic of Korea.,Lung Cancer Center, Kyungpook National University Medical Center, Daegu, Republic of Korea
| | - Mi Jeong Hong
- Department of Biochemistry and Department of Cell Biology, Kyungpook National University, Daegu, Republic of Korea.,BK21 Plus KNU Biomedical Convergence Program, Department of Biomedical Science, Kyungpook National University, Daegu, Republic of Korea
| | - Hyo-Sung Jeon
- Department of Biochemistry and Department of Cell Biology, Kyungpook National University, Daegu, Republic of Korea
| | - Yi Young Choi
- Department of Biochemistry and Department of Cell Biology, Kyungpook National University, Daegu, Republic of Korea
| | - Jin Eun Choi
- Department of Biochemistry and Department of Cell Biology, Kyungpook National University, Daegu, Republic of Korea.,Cell and Matrix Research Institute, Kyungpook National University, Daegu, Republic of Korea
| | - Hyo-Gyoung Kang
- Department of Biochemistry and Department of Cell Biology, Kyungpook National University, Daegu, Republic of Korea.,Cell and Matrix Research Institute, Kyungpook National University, Daegu, Republic of Korea
| | - Deuk Kju Jung
- Department of Biochemistry and Department of Cell Biology, Kyungpook National University, Daegu, Republic of Korea.,BK21 Plus KNU Biomedical Convergence Program, Department of Biomedical Science, Kyungpook National University, Daegu, Republic of Korea
| | - Chengcheng Jin
- Department of Biochemistry and Department of Cell Biology, Kyungpook National University, Daegu, Republic of Korea.,BK21 Plus KNU Biomedical Convergence Program, Department of Biomedical Science, Kyungpook National University, Daegu, Republic of Korea
| | - Sook Kyung Do
- Department of Biochemistry and Department of Cell Biology, Kyungpook National University, Daegu, Republic of Korea.,BK21 Plus KNU Biomedical Convergence Program, Department of Biomedical Science, Kyungpook National University, Daegu, Republic of Korea
| | - Seung Soo Yoo
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Daegu, Republic of Korea.,Lung Cancer Center, Kyungpook National University Medical Center, Daegu, Republic of Korea
| | - Yangki Seok
- Thoracic Surgery, Kyungpook National University, Daegu, Republic of Korea
| | - Eung Bae Lee
- Thoracic Surgery, Kyungpook National University, Daegu, Republic of Korea
| | - Kyung Min Shin
- Radiology, Kyungpook National University, Daegu, Republic of Korea
| | - Ji Yun Jeong
- Pathology, Kyungpook National University, Daegu, Republic of Korea
| | - Won Kee Lee
- Biostatistics Center, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Jaehee Lee
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Seung Ick Cha
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Chang Ho Kim
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Young Tae Kim
- Department of Thoracic and Cardiovascular Surgery, Seoul National University School of Medicine, Seoul, Republic of Korea
| | - Sanghoon Jheon
- Department of Thoracic and Cardiovascular Surgery, Seoul National University School of Medicine, Seoul, Republic of Korea
| | - Jae Yong Park
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Daegu, Republic of Korea.,Lung Cancer Center, Kyungpook National University Medical Center, Daegu, Republic of Korea.,Department of Biochemistry and Department of Cell Biology, Kyungpook National University, Daegu, Republic of Korea.,Cell and Matrix Research Institute, Kyungpook National University, Daegu, Republic of Korea.,BK21 Plus KNU Biomedical Convergence Program, Department of Biomedical Science, Kyungpook National University, Daegu, Republic of Korea
| |
Collapse
|
23
|
Geng P, Ou J, Li J, Liao Y, Wang N, Xie G, Sa R, Liu C, Xiang L, Liang H. A Comprehensive Analysis of Influence ERCC Polymorphisms Confer on the Development of Brain Tumors. Mol Neurobiol 2015; 53:2705-14. [PMID: 26264164 DOI: 10.1007/s12035-015-9371-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2014] [Accepted: 07/21/2015] [Indexed: 12/21/2022]
Abstract
Within DNA repair genes, there lie a number of single nucleotide polymorphisms that may impair protein function and attenuate DNA repair capability, resulting in genomic instability and individual predisposition to malignancies. The purpose of this study was to assess the previously reported inconsistent association of polymorphisms in ERCC1 (rs11615, rs3212986), ERCC2 (rs13181, rs1799793, rs238406), and ERCC5 (rs17655) with the development of brain tumors. In the present work, we carried out a comprehensive meta-analysis of results from all published data (5 data sets for rs11615, 7 for rs3212986, 11 for rs13181, 5 for rs1799793, 3 for rs238406, and 4 for rs17655) to evaluate risk of brain tumors contributed by the polymorphisms being investigated. Either the analytic method described by Mantel and Haenszel or that proposed by DerSimonian and Laird was properly used to summarize the risk estimates (OR and 95% CI). Data analyses were done with Stata version 12.0. Meta-analyses were performed for all polymorphisms, and only rs3212986 in the ERCC1 gene showed a significant association with glioma incidence. In the homozygote comparison, we found 1.26-fold elevated risk of glioma in relation to presence of the AA genotype (OR = 1.26, 95% CI = 1.05-1.52, P OR = 0.013, P heterogeneity = 0.849, I(2) = 0.0%). We also noted that individuals with the rs3212986-AA as compared to those with rs3212986-CC/CA had a 28% higher risk to develop glioma (OR = 1.28, 95% CI = 1.06-1.53, P OR = 0.008, Pheterogeneity = 0.808, I(2) = 0.0%). No major effects were observed for Caucasians or Asians in subgroup analysis by ethnicity. ERCC1 rs3212986 is a common single nucleotide polymorphism and may contribute toward individual susceptibility for glioma. Further research in this filed is required to verify the association obtained based on a relatively small number.
Collapse
Affiliation(s)
- Peiliang Geng
- Department of Oncology and Southwest Cancer Center, Southwest Hospital Third Military Medical University, 29 Gaotanyan Main Street, Chongqing, 400038, China
| | - Juanjuan Ou
- Department of Oncology and Southwest Cancer Center, Southwest Hospital Third Military Medical University, 29 Gaotanyan Main Street, Chongqing, 400038, China
| | - Jianjun Li
- Department of Oncology and Southwest Cancer Center, Southwest Hospital Third Military Medical University, 29 Gaotanyan Main Street, Chongqing, 400038, China
| | - Yunmei Liao
- Department of Oncology and Southwest Cancer Center, Southwest Hospital Third Military Medical University, 29 Gaotanyan Main Street, Chongqing, 400038, China
| | - Ning Wang
- Department of Oncology and Southwest Cancer Center, Southwest Hospital Third Military Medical University, 29 Gaotanyan Main Street, Chongqing, 400038, China
| | - Ganfeng Xie
- Department of Oncology and Southwest Cancer Center, Southwest Hospital Third Military Medical University, 29 Gaotanyan Main Street, Chongqing, 400038, China
| | - Rina Sa
- Department of Oncology and Southwest Cancer Center, Southwest Hospital Third Military Medical University, 29 Gaotanyan Main Street, Chongqing, 400038, China
| | - Chen Liu
- Department of Oncology and Southwest Cancer Center, Southwest Hospital Third Military Medical University, 29 Gaotanyan Main Street, Chongqing, 400038, China
| | - Lisha Xiang
- Department of Oncology and Southwest Cancer Center, Southwest Hospital Third Military Medical University, 29 Gaotanyan Main Street, Chongqing, 400038, China
| | - Houjie Liang
- Department of Oncology and Southwest Cancer Center, Southwest Hospital Third Military Medical University, 29 Gaotanyan Main Street, Chongqing, 400038, China.
| |
Collapse
|
24
|
Cui QK, Zhu JX, Liu WD, Wang YH, Wang ZG. Association of ERCC1 rs3212986 & ERCC2 rs13181 polymorphisms with the risk of glioma. Pak J Med Sci 2015; 30:1409-14. [PMID: 25674148 PMCID: PMC4320740 DOI: 10.12669/pjms.306.5221] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2014] [Revised: 07/16/2014] [Accepted: 08/04/2014] [Indexed: 11/15/2022] Open
Abstract
Objective:: Several previous studies have reported the role variant of ERCC1 rs3212986 and ERCC2 rs13181 polymorphisms in the risk of glioma, but the results of these studies are inconsistent. Therefore, we aimed to conduct a meta-analysis to investigate the role of ERCC1 rs3212986 and ERCC2 rs13181 on the risk of glioma. Methods: A comprehensive research was conducted through the databases of Pubmed, EMBASE and the China National Knowledge Infrastructure (CNKI) platforms until June 1, 2014, including 14 eligible case-control studies. Results: Our meta-analysis found that ERCC1 rs3212986 AA genotype was significantly associated with increased risk of glioma compared with CC genotype, and the pooled OR (95%CI) was 1.29(1.07-1.55). By subgroup analysis, ERCC1 rs3212986 AA genotype was found to be significantly correlated with increased glioma risk in Chinese population (OR=1.37, 95%CI=1.07, 1.55), Similarly, we found that ERCC2 rs13181 GT and TT genotypes were significantly associated with increased risk of glioma in Chinese population, with ORs(95%CI) of 1.47(1.17-1.85) and 1.50(1.02-2.22). But ERCC1 rs3212986 and ERCC2 rs13181 polymorphisms had no significant association with glioma risk in Caucasian populations. By begg’s funnel plot, we found that no publication bias was existed in this meta-analysis. Conclusion: Our meta-analysis suggested that ERCC1 rs3212986 and ERCC2 rs13181 polymorphism play an important risk factor for brain tumor development in Chinese population, but no association in Caucasian populations.
Collapse
Affiliation(s)
- Qing-Ke Cui
- Qing-ke Cui, Department of Neurosurgery, Liaocheng People's Hospital, Liaocheng Shandong, 252000, P. R. China
| | - Jian-Xin Zhu
- Qing-ke Cui, Department of Neurosurgery, Liaocheng People's Hospital, Liaocheng Shandong, 252000, P. R. China
| | - Wei-Dong Liu
- Wei-dong Liu, Department of Neurosurgery, Liaocheng People's Hospital, Liaocheng Shandong, 252000, P. R. China
| | - Yun-Hua Wang
- Yun-hua Wang, Department of Neurosurgery, Liaocheng People's Hospital, Liaocheng Shandong, 252000, P. R. China
| | - Zhi-Gang Wang
- Zhi-gang Wang, Department of Neurosurgery, Liaocheng People's Hospital, Liaocheng Shandong, 252000, P. R. China
| |
Collapse
|
25
|
He LW, Shi R, Jiang L, Zeng Y, Ma WL, Zhou JY. XRCC1 gene polymorphisms and glioma risk in Chinese population: a meta-analysis. PLoS One 2014; 9:e111981. [PMID: 25375625 PMCID: PMC4222958 DOI: 10.1371/journal.pone.0111981] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2014] [Accepted: 10/03/2014] [Indexed: 01/18/2023] Open
Abstract
Background Three extensively investigated polymorphisms (Arg399Gln, Arg194Trp, and Arg280His) in the X-ray repair cross-complementing group 1 (XRCC1) gene have been implicated in risk for glioma. However, the results from different studies remain inconsistent. To clarify these conflicts, we performed a quantitative synthesis of the evidence to elucidate these associations in the Chinese population. Methods Data were extracted from PubMed and EMBASE, with the last search up to August 21, 2014. Meta-analysis was performed by critically reviewing 8 studies for Arg399Gln (3062 cases and 3362 controls), 8 studies for Arg194Trp (3419 cases and 3680 controls), and 5 studies for Arg280His (2234 cases and 2380 controls). All of the statistical analyses were performed using the software program, STATA (version 11.0). Results Our analysis suggested that both Arg399Gln and Arg194Trp polymorphisms were significantly associated with increased risk of glioma (for Arg399Gln polymorphism: Gln/Gln vs. Arg/Arg, OR = 1.82, 95% CI = 1.46–2.27, P = 0.000; Arg/Gln vs. Arg/Arg, OR = 1.25, 95% CI = 1.10–1.42, P = 0.001 and for Arg194Trp polymorphism: recessive model, OR = 1.78, 95% CI = 1.44–2.19, P = 0.000), whereas the Arg280His polymorphism had no influence on the susceptibility to glioma in a Chinese population. Conclusions This meta-analysis suggests that there may be no association between the Arg280His polymorphism and glioma risk, whereas the Arg399Gln/Arg194Trp polymorphisms may contribute to genetic susceptibility to glioma in the Chinese population. Nevertheless, large-scale, well-designed and population-based studies are needed to further evaluate gene-gene and gene–environment interactions, as well as to measure the combined effects of these XRCC1 variants on glioma risk.
Collapse
Affiliation(s)
- Li-Wen He
- Institute of Genetic Engineering, Southern Medical University, Guangzhou, China
- Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Rong Shi
- Institute of Genetic Engineering, Southern Medical University, Guangzhou, China
| | - Lei Jiang
- Department of Neurosurgery, Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Ye Zeng
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Wen-Li Ma
- Institute of Genetic Engineering, Southern Medical University, Guangzhou, China
| | - Jue-Yu Zhou
- Institute of Genetic Engineering, Southern Medical University, Guangzhou, China
- * E-mail:
| |
Collapse
|